551
|
Singanayagam A, Triantafyllou E. Macrophages in Chronic Liver Failure: Diversity, Plasticity and Therapeutic Targeting. Front Immunol 2021; 12:661182. [PMID: 33868313 PMCID: PMC8051585 DOI: 10.3389/fimmu.2021.661182] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic liver injury results in immune-driven progressive fibrosis, with risk of cirrhosis development and impact on morbidity and mortality. Persistent liver cell damage and death causes immune cell activation and inflammation. Patients with advanced cirrhosis additionally experience pathological bacterial translocation, exposure to microbial products and chronic engagement of the immune system. Bacterial infections have a high incidence in cirrhosis, with spontaneous bacterial peritonitis being the most common, while the subsequent systemic inflammation, organ failure and immune dysregulation increase the mortality risk. Tissue-resident and recruited macrophages play a central part in the development of inflammation and fibrosis progression. In the liver, adipose tissue, peritoneum and intestines, diverse macrophage populations exhibit great phenotypic and functional plasticity determined by their ontogeny, epigenetic programming and local microenvironment. These changes can, at different times, promote or ameliorate disease states and therefore represent potential targets for macrophage-directed therapies. In this review, we discuss the evidence for macrophage phenotypic and functional alterations in tissue compartments during the development and progression of chronic liver failure in different aetiologies and highlight the potential of macrophage modulation as a therapeutic strategy for liver disease.
Collapse
Affiliation(s)
- Arjuna Singanayagam
- Infection and Immunity Clinical Academic Group, St. George’s University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
552
|
Wan M, Han J, Ding L, Hu F, Gao P. Novel Immune Subsets and Related Cytokines: Emerging Players in the Progression of Liver Fibrosis. Front Med (Lausanne) 2021; 8:604894. [PMID: 33869241 PMCID: PMC8047058 DOI: 10.3389/fmed.2021.604894] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a pathological process caused by persistent chronic injury of the liver. Kupffer cells, natural killer (NK) cells, NKT cells, and dendritic cells (DCs), which are in close contact with T and B cells, serve to bridge innate and adaptive immunity in the liver. Meanwhile, an imbalanced inflammatory response constitutes a challenge in liver disease. The dichotomous roles of novel immune cells, including T helper 17 (Th17), regulatory T cells (Tregs), mucosa-associated invariant T cells (MAIT), and innate lymphoid cells (ILCs) in liver fibrosis have gradually been revealed. These cells not only induce damage during liver fibrosis but also promote tissue repair. Hence, immune cells have unique, and often opposing, roles during the various stages of fibrosis. Due to this heterogeneity, the treatment, or reversal of fibrosis through the target of immune cells have attracted much attention. Moreover, activation of hepatic stellate cells (HSCs) constitutes the core of fibrosis. This activation is regulated by various immune mediators, including Th17, Th22, and Th9, MAIT, ILCs, and γδ T cells, as well as their related cytokines. Thus, liver fibrosis results from the complex interaction of these immune mediators, thereby complicating the ability to elucidate the mechanisms of action elicited by each cell type. Future developments in biotechnology will certainly aid in this feat to inform the design of novel therapeutic targets. Therefore, the aim of this review was to summarize the role of specific immune cells in liver fibrosis, as well as biomarkers and treatment methods related to these cells.
Collapse
Affiliation(s)
- Minjie Wan
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China.,Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jiawen Han
- Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Lili Ding
- Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China.,Intensive Care Unit, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Feng Hu
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital, Jilin University, Changchun, China
| | - Pujun Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
553
|
Zhang P, Gan Z, Tang L, Zhou L, Huang X, Wang J. WITHDRAWN: Exosomes from microRNA-145-5p-modified HUCB-MSCs attenuate CCl4-induced hepatic fibrosis via down-regulating FSCN1 expression. Life Sci 2021:119404. [PMID: 33794251 DOI: 10.1016/j.lfs.2021.119404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Pan Zhang
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Zeying Gan
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Lanyan Tang
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Lizhi Zhou
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Xin Huang
- Department of Infection, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China
| | - Jianlong Wang
- Department of Orthopedics, No.3 Hospital of Xiangya, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
554
|
de Moraes ACN, de Andrade CBV, Ramos IPR, Dias ML, Batista CMP, Pimentel CF, de Carvalho JJ, Goldenberg RCDS. Resveratrol promotes liver regeneration in drug-induced liver disease in mice. Food Res Int 2021; 142:110185. [PMID: 33773662 DOI: 10.1016/j.foodres.2021.110185] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 01/20/2021] [Accepted: 01/24/2021] [Indexed: 12/12/2022]
Abstract
Studies suggest that the bioactive polyphenolic compound resveratrol (RESV, trans-isomer), found naturally in certain foods such as red grapes and peanuts, may be able to ameliorate liver damage. However, the effects and efficacy of long-term treatment with RESV remain unclear. Here, we used an acetaminophen (APAP; 400 mg/kg/d for 15 days) overdose model to induce liver damage in C56BL/6 mice. Three days after the intoxication was stopped, we observed biochemical, histological and ultrastructural alterations in the livers of these mice. The APAP-treated animals were then given RESV (10 mg/kg/d) for 60 days. Blood and tissue were analyzed at days 7, 30 and 60. Our data show that long-term RESV treatment (60 days) ameliorates the liver injury caused by APAP intoxication, restoring histological features, ultrastructural organization and serum biochemical parameters (albumin, alanine aminotransferase). Ck18- and F4/80-positive cells (indicators of hepatocyte recovery) were reestablished and the number of α-SMA positive cells was normalized after long-term RESV treatment. Additionally, downregulation of the drug transporter BCRP was observed. Electron microscopy revealed that treatment with RESV was effective in restoring the shape and size of hepatic microvilli and normalizing both the number and viability of mitochondria. Taken together, these results indicate that long-term treatment with RESV is effective in alleviating liver injury caused by APAP administration.
Collapse
Affiliation(s)
- Alan Cesar Nunes de Moraes
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil; Biology Department, Federal Fluminense University, UFF, Niterói, RJ, Brazil
| | - Cherley Borba Vieira de Andrade
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil; Department of Histology and Embryology, State University of Rio de Janeiro, UERJ, Rio de Janeiro, RJ, Brazil
| | - Isalira Peroba Rezende Ramos
- Center for Structural Biology and Bio-imaging, CENABIO, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, Brazil
| | - Marlon Lemos Dias
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Cintia Marina Paz Batista
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Cibele Ferreira Pimentel
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Jorge Jose de Carvalho
- Department of Histology and Embryology, State University of Rio de Janeiro, UERJ, Rio de Janeiro, RJ, Brazil
| | - Regina Coeli Dos Santos Goldenberg
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, INCT-REGENERA, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.
| |
Collapse
|
555
|
Tangphokhanon W, Pradidarcheep W, Lametschwandtner A. α-mangostin preserves hepatic microvascular architecture in fibrotic rats as shown by scanning electron microscopy of vascular corrosion casts. Biomed Rep 2021; 14:48. [PMID: 33859819 PMCID: PMC8042669 DOI: 10.3892/br.2021.1424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/04/2021] [Indexed: 11/17/2022] Open
Abstract
Liver fibrosis is a dynamic condition caused by wound-healing in which scar tissue replaces the liver parenchyma following repetitive injuries. It is hypothesized that α-mangostin (AM), the major constituent of the xanthone fraction in extracts of Garcinia mangostana L., may protect the hepatic microvascular bed from thioacetamide (TAA)-induced fibrosis. In the present study, rats were divided into 4 groups: Control rats received no treatment; TAA-treated rats received 150 mg/kg TAA 3 times per week intraperitoneally; AM-treated rats received 75 mg/kg AM twice per week intraperitoneally; and TAA+AM-treated rats received both TAA and AM as described above. Rat livers were processed either for light microscopy or for vascular corrosion casting after 30 and 60 days of treatment. Vascular parameters were measured by 3D morphometry analysis of scanning electron micrographs. AM attenuated hepatocellular injuries and delayed both periportal and pericentral fibrosis in the TAA-treated rats. The comparison of findings at day 30 and 60 showed that TAA-induced fibrotic changes were progressive in time, and that the beneficial effects of AM only became apparent after prolonged treatment. The livers of rats treated with both TAA and AM had less space surrounding the portal vessels, improved preservation of the hepatic microvascular pattern, and minimally altered sinusoidal patterns with few signs of terminal portal venule remodeling. AM therefore partially protected the liver against hepatotoxin-induced fibrosis and the associated microvascular changes. The mechanism of the protective effect of AM on the liver remains to be investigated.
Collapse
Affiliation(s)
- Wasan Tangphokhanon
- Center of Excellence in Veterinary Biosciences, Department of Veterinary Biosciences and Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Wisuit Pradidarcheep
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Alois Lametschwandtner
- Department of Biosciences, Vascular and Exercise Biology Unit, University of Salzburg, Salzburg 5020, Austria
| |
Collapse
|
556
|
He C, Shu B, Zhou Y, Zhang R, Yang X. The miR-139-5p/peripheral myelin protein 22 axis modulates TGF-β-induced hepatic stellate cell activation and CCl 4-induced hepatic fibrosis in mice. Life Sci 2021; 276:119294. [PMID: 33675896 DOI: 10.1016/j.lfs.2021.119294] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022]
Abstract
Hepatic stellate cells (HSCs) are the major source of extracellular matrix (ECM)-producing myofibroblasts. When activated by multiple injuries, HSCs become proliferative, contractile, inflammatory and chemotactic and are characterized by enhanced ECM production, which plays a central role in hepatic fibrosis initiation and progression. In the present study, through bioinformatics analysis, we identified the abnormal upregulation of Peripheral Myelin Protein 22 (PMP22) in fibrotic murine liver. In CCl4-induced hepatic fibrosis model in mice and TGF-β-activated hHSCs, PMP22 was observed remarkably upregulated. In TGF-β-stimulated hHSCs, PMP22 silencing hindered, whereas PMP22 overexpression aggravated TGF-β-induced hHSC activation. In CCl4-induced hepatic fibrosis model in mice, PMP22 silencing improved CCl4-caused liver damage and fibrotic changes. Through online tools prediction and experimental validation, miR-139-5p was found to bind to the 3'UTR of PMP22 and negatively regulate the expression of PMP22. In contrast to PMP22 silencing, miR-139-5p inhibition enhanced TGF-β-induced hHSC activation; the effects of miR-139-5p inhibition on TGF-β-induced hHSC activation were partially reversed by PMP22 silencing. In conclusion, we identify the abnormal upregulation of PMP22 in TGF-β-activated HSCs and CCl4-induced hepatic fibrosis model in mice, as well as the pro-fibrotic role of PMP22 through aggravating TGF-β-induced HSCs activation. miR-139-5p targets the 3'UTR of PMP22 and inhibits PMP22 expression; miR-139-5p hinders TGF-β-induced HSCs activation through targeting PMP22.
Collapse
Affiliation(s)
- Chao He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Bo Shu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yingxia Zhou
- Department of Surgical Operation, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Ruizhi Zhang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xin Yang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
557
|
Li H. Angiogenesis in the progression from liver fibrosis to cirrhosis and hepatocelluar carcinoma. Expert Rev Gastroenterol Hepatol 2021; 15:217-233. [PMID: 33131349 DOI: 10.1080/17474124.2021.1842732] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Persistent inflammation and hypoxia are strong stimulus for pathological angiogenesis and vascular remodeling, and are also the most important elements resulting in liver fibrosis. Sustained inflammatory process stimulates fibrosis to the end-point of cirrhosis and sinusoidal portal hypertension is an important feature of cirrhosis. Neovascularization plays a pivotal role in collateral circulation formation of portal vein, mesenteric congestion, and high perfusion. Imbalance of hepatic artery and portal vein blood flow leads to the increase of hepatic artery inflow, which is beneficial to the formation of nodules. Angiogenesis contributes to progression from liver fibrosis to cirrhosis and hepatocellular carcinoma (HCC) and anti-angiogenesis therapy can improve liver fibrosis, reduce portal pressure, and prolong overall survival of patients with HCC. Areas covers: This paper will try to address the difference of the morphological characteristics and mechanisms of neovascularization in the process from liver fibrosis to cirrhosis and HCC and further compare the different efficacy of anti-angiogenesis therapy in these three stages. Expert opinion: More in-depth understanding of the role of angiogenesis factors and the relationship between angiogenesis and other aspects of the pathogenesis and transformation may be the key to enabling future progress in the treatment of patients with liver fibrosis, cirrhosis, and HCC.
Collapse
Affiliation(s)
- Hui Li
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine , Chengdu, Sichuan Province, P. R. China
| |
Collapse
|
558
|
Zhou X, Rao J, Wu X, Deng R, Ma Y. Comparison of 2-D Shear Wave Elastography and Point Shear Wave Elastography for Assessing Liver Fibrosis. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:408-427. [PMID: 33342618 DOI: 10.1016/j.ultrasmedbio.2020.11.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/08/2020] [Accepted: 11/16/2020] [Indexed: 06/12/2023]
Abstract
Progressive liver fibrosis may result in cirrhosis, portal hypertension and increased risk of hepatocellular carcinoma. We performed a meta-analysis to compare liver fibrosis staging in chronic liver disease patients using 2-D shear wave elastography (2-D SWE) and point shear wave elastography (pSWE). The PubMed, Web of Science and Cochrane Library databases were searched until May 31, 2020 for studies evaluating the diagnostic performance of 2-D SWE and pSWE in assessing liver fibrosis. Pooled sensitivity, specificity, positive and negative likelihood ratios, diagnostic odds ratios and area under receiver operating characteristic curve were estimated using the bivariate random effects model. As a result, 71 studies with 11,345 patients were included in the analysis. The pooled sensitivities of 2-D SWE and pSWE significantly differed for the detection of significant fibrosis (F ≥ 2; 0.84 vs. 0.76, p < 0.001) and advanced fibrosis (F ≥ 3; 0.90 vs. 0.83, p = 0.003), but not for detection of cirrhosis (F = 4; 0.89 vs. 0.85, p = 0.090). The pooled specificities of 2-D SWE and pSWE did not significantly differ for detection of F ≥ 2 (0.81 vs. 0.79, p = 0.753), F ≥ 3 (0.87 vs. 0.83, p = 0.163) or F = 4 (0.87 vs. 0.84, p = 0.294). Both 2-D SWE and pSWE have high sensitivity and specificity for detecting each stage of liver fibrosis. Two-dimensional SWE has higher sensitivity than pSWE for detection of significant fibrosis and advanced fibrosis.
Collapse
Affiliation(s)
- Xiaozhuan Zhou
- Organ Transplant Center, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiawei Rao
- Organ Transplant Center, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xukun Wu
- Organ Transplant Center, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ronghai Deng
- Organ Transplant Center, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yi Ma
- Organ Transplant Center, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
559
|
Trebicka J, Bork P, Krag A, Arumugam M. Utilizing the gut microbiome in decompensated cirrhosis and acute-on-chronic liver failure. Nat Rev Gastroenterol Hepatol 2021; 18:167-180. [PMID: 33257833 DOI: 10.1038/s41575-020-00376-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
The human gut microbiome has emerged as a major player in human health and disease. The liver, as the first organ to encounter microbial products that cross the gut epithelial barrier, is affected by the gut microbiome in many ways. Thus, the gut microbiome might play a major part in the development of liver diseases. The common end stage of liver disease is decompensated cirrhosis and the further development towards acute-on-chronic liver failure (ACLF). These conditions have high short-term mortality. There is evidence that translocation of components of the gut microbiota, facilitated by different pathogenic mechanisms such as increased gut epithelial permeability and portal hypertension, is an important driver of decompensation by induction of systemic inflammation, and thereby also ACLF. Elucidating the role of the gut microbiome in the aetiology of decompensated cirrhosis and ACLF deserves further investigation and improvement; and might be the basis for development of diagnostic and therapeutic strategies. In this Review, we focus on the possible pathogenic, diagnostic and therapeutic role of the gut microbiome in decompensation of cirrhosis and progression to ACLF.
Collapse
Affiliation(s)
- Jonel Trebicka
- Translational Hepatology, Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany. .,European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain. .,Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark. .,Institute for Bioengineering of Catalonia, Barcelona, Spain.
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Aleksander Krag
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Manimozhiyan Arumugam
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark. .,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
560
|
Gabbiadini R, Zacharopoulou E, Furfaro F, Craviotto V, Zilli A, Gilardi D, Roda G, Loy L, Fiorino G, Peyrin-Biroulet L, Danese S, Allocca M. Application of Ultrasound Elastography for Assessing Intestinal Fibrosis in Inflammatory Bowel Disease: Fiction or Reality? Curr Drug Targets 2021; 22:347-355. [PMID: 33213341 DOI: 10.2174/1389450121666201119142919] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/07/2020] [Accepted: 09/27/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Intestinal fibrosis and subsequent strictures represent an important burden in inflammatory bowel disease (IBD). Both the detection and evaluation of the degree of fibrosis in stricturing Crohn's disease (CD) are important when deciding the best therapeutic strategy (medical anti-inflammatory therapy, endoscopic dilation, surgery). Ultrasound elastography (USE) is a non-invasive technique that has been proposed in the field of IBD for evaluating intestinal stiffness as a biomarker of intestinal fibrosis. OBJECTIVE The aim of this review is to discuss the ability and current role of ultrasound elastography in the assessment of intestinal fibrosis. RESULTS AND CONCLUSION Data on USE in IBD are provided by pilot and proof-of-concept studies with small sample size. The first type of USE investigated was strain elastography, while shear wave elastography has been introduced recently. Despite the heterogeneity of the methods of the studies, USE has been proven to be able to assess intestinal fibrosis in patients with stricturing CD. However, before introducing this technique in current practice, further studies with larger sample sizes are needed. In addition, the use of homogeneous parameters, the assessment of reproducibility, and the identification of validated cut-off values are essential.
Collapse
Affiliation(s)
- Roberto Gabbiadini
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Eirini Zacharopoulou
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Federica Furfaro
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Vincenzo Craviotto
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Alessandra Zilli
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Daniela Gilardi
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Giulia Roda
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Laura Loy
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Gionata Fiorino
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology and Inserm U954, University Hospital of Nancy, Lorraine University, Nancy, France
| | - Silvio Danese
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| | - Mariangela Allocca
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano (Mi), Italy
| |
Collapse
|
561
|
Lambrecht J, Tacke F. Controversies and Opportunities in the Use of Inflammatory Markers for Diagnosis or Risk Prediction in Fatty Liver Disease. Front Immunol 2021; 11:634409. [PMID: 33633748 PMCID: PMC7900147 DOI: 10.3389/fimmu.2020.634409] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022] Open
Abstract
In the Western society, non-alcoholic fatty liver disease (NAFLD), characterized by the excessive accumulation of fat in the liver, represents the most common cause of chronic liver disease. If left untreated, approximately 15%-20% of patients with NAFLD will progress to non-alcoholic steatohepatitis (NASH), in which lobular inflammation, hepatocyte ballooning and fibrogenesis further contribute to a distorted liver architecture and function. NASH initiation has significant effects on liver-related mortality, as even the presence of early stage fibrosis increases the chances of adverse patient outcome. Therefore, adequate diagnostic tools for NASH are needed, to ensure that relevant therapeutic actions can be taken as soon as necessary. To date, the diagnostic gold standard remains the invasive liver biopsy, which is associated with several drawbacks such as high financial costs, procedural risks, and inter/intra-observer variability in histology analysis. As liver inflammation is a major hallmark of disease progression, inflammation-related circulating markers may represent an interesting source of non-invasive biomarkers for NAFLD/NASH. Examples for such markers include cytokines, chemokines or shed receptors from immune cells, circulating exosomes related to inflammation, and changing proportions of peripheral blood mononuclear cell (PBMC) subtypes. This review aims at documenting and critically discussing the utility of such novel inflammatory markers for NAFLD/NASH-diagnosis, patient stratification and risk prediction.
Collapse
Affiliation(s)
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
562
|
Huang W, Huang F, Zhang R, Luo H. LncRNA Neat1 expedites the progression of liver fibrosis in mice through targeting miR-148a-3p and miR-22-3p to upregulate Cyth3. Cell Cycle 2021; 20:490-507. [PMID: 33550894 DOI: 10.1080/15384101.2021.1875665] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Liver fibrosis is a common response to chronic liver injury, ultimately leading to cirrhosis. The activation of hepatic stellate cells (HSCs) plays a dominant role in liver fibrosis. The regulatory roles of long noncoding RNAs (lncRNAs) in multiple human diseases have been observed. This study was dedicated to investigating the regulatory effects of the lncRNA nuclear paraspeckle assembly transcript 1 (Neat1) on liver fibrosis and HSC activation. Upregulation of Neat1 and cytohesin 3 (Cyth3) and downregulation of miR-148a-3p and miR-22-3p were observed in mouse fibrotic liver tissues. Knockdown of Neat1 or Cyth3 attenuated liver fibrosis and collagen deposition in vivo and the activation of HSCs in vitro. An miR-148a-3p and miR-22-3p inhibitor facilitated HSC activation and collagen fiber expression. Neat1 directly targeted miR-148a-3p and miR-22-3p to modulate Cyth3 expression. Knockdown of Neat1 inhibited Cyth3 expression via the competing endogenous RNA (ceRNA) mechanism of sponging miR-148a-3p and miR-22-3p to regulate liver fibrosis and HSC activation. The ceRNA regulatory network may promote a better understanding of liver fibrogenesis, contribute to an original agreement of liver fibrosis etiopathogenesis and provide insights into the development of a novel domain of lncRNA-directed therapy against liver fibrosis.
Collapse
Affiliation(s)
- Wei Huang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Feizhou Huang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Rui Zhang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongwu Luo
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
563
|
Kuang M, Wu H, Hu L, Guo X, He D, Liu B, Chen M, Gu J, Gu J, Zeng X, Ruan Y. Up-regulation of FUT8 inhibits TGF-β1-induced activation of hepatic stellate cells during liver fibrogenesis. Glycoconj J 2021; 38:77-87. [PMID: 33608773 DOI: 10.1007/s10719-021-09975-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/28/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
Liver fibrosis is a continuous wound healing response caused by chronic liver injury, and the activation of hepatic stellate cells (HSCs) is considered as the main event for it. Core fucosylation catalyzed by FUT8 refers to adding the fucosyl moiety to the innermost GlcNAc residue of N-linked oligosaccharides and is involved in many biological processes such as cell differentiation, migration, and signaling transduction. Aberrant core fucosylation is associated with a variety of diseases including cardiovascular disease, tumors and neuroinflammation, but much less is understood in liver fibrosis. Herein, we reported FUT8 mRNA level was increased in patients with liver fibrosis from GEO database and positively correlated with fibrosis progression. FUT8 expression and the core fucosylation were also elevated in TAA-induced mouse liver fibrosis model, and were mainly distributed in the fibrous septum of mouse liver. TGF-β1, as the most pro-fibrogenic cytokine, could promote the expression of FUT8 and total core fucosylation levels in HSCs in vitro. However, up-regulation of FUT8 in turn inhibited TGF-β1-induced trans-differentiation, migration and pro-fibrogenic signaling pathways in HSCs. In conclusion, our results suggest that the up-regulation of FUT8 inhibits TGF-β1-induced HSC activation in a negative feedback loop, and provide potential new therapeutic strategy for liver fibrosis by targeting FUT8.
Collapse
Affiliation(s)
- Mengzhen Kuang
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Hao Wu
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Lan Hu
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xinying Guo
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Daochuan He
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Bo Liu
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Mengqian Chen
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jie Gu
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jianxin Gu
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaoqing Zeng
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Yuanyuan Ruan
- NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
564
|
Shu Y, Liu X, Huang H, Wen Q, Shu J. Research progress of natural compounds in anti-liver fibrosis by affecting autophagy of hepatic stellate cells. Mol Biol Rep 2021; 48:1915-1924. [PMID: 33609264 PMCID: PMC7925445 DOI: 10.1007/s11033-021-06171-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022]
Abstract
Chronic liver diseases caused by various pathogenesis are marked by inflammatory infiltration and wound healing reaction, while their normal regeneration ability is impaired. The unbalance between the generation and the degradation of extracellular matrix (ECM) leads to collagen accumulation and develops into liver fibrosis. Inflammation, oxidative stress, and autophagy interact closely in the pathogenesis of hepatic fibrosis. Reactive Oxygen Species (ROS) can not only stimulate Kupffer cells to release massive inflammatory factors, but induce autophagy. However, the latter may suppress inflammatory reaction by inhibiting proinflammatory complex formation directly, and removing damaged organelles or pathogenic microorganism indirectly. At present, effective anti-fibrosis drugs are still lacking. Previous studies have found various natural compounds enabled liver protection through anti-inflammatory, antioxidant, and other mechanisms. In recent years, autophagy, a vital life activity, has been found to be involved in the mechanism of liver fibrosis. As a new target, developing anti-liver fibrosis drugs that regulate the activity of autophagy is very promising. In this review, we summarize the latest studies about natural compounds in the treatment of liver fibrosis by regulating autophagy.
Collapse
Affiliation(s)
- Yongxiang Shu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Xuyou Liu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Haifeng Huang
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Qi Wen
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| | - Jianchang Shu
- Department of Gastroenterology, GuangZhou Red Cross Hospital, Jinan University, Guangzhou, 510220 China
| |
Collapse
|
565
|
Apigenin Alleviates Liver Fibrosis by Inhibiting Hepatic Stellate Cell Activation and Autophagy via TGF- β1/Smad3 and p38/PPAR α Pathways. PPAR Res 2021; 2021:6651839. [PMID: 33574836 PMCID: PMC7861947 DOI: 10.1155/2021/6651839] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/10/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
Objective The aim of this study is to confirm the hepatocellular protective functions of apigenin and the molecular mechanism on liver fibrosis in mice. Methods Carbon tetrachloride (CCl4) and bile duct ligature (BDL) mouse fibrosis models were used to investigate the effects of apigenin on liver fibrosis. Sixty-six male C57 mice were randomly divided into eight groups, including the vehicle group, CCl4 group, CCl4+L-apigenin (20 mg/kg) group, CCl4+H-apigenin (40 mg/kg) group, sham group, BDL group, BDL+L-apigenin(20 mg/kg) group, and BDL+H-apigenin(40 mg/kg) group. Serum liver enzymes (ALT and AST), proteins associated with autophagy, and indicators linked with the TGF-β1/Smad3 and p38/PPARα pathways were detected using qRT-PCR, immunohistochemical staining, and western blotting. Results Our findings confirmed that apigenin could decrease the levels of ALT and AST, suppress the generation of ECM, inhibit the activation of HSCs, regulate the balance of MMP2 and TIMP1, reduce the expression of autophagy-linked protein, and restrain the TGF-β1/Smad3 and p38/PPARα pathways. Conclusion Apigenin could alleviate liver fibrosis by inhibiting hepatic stellate cell activation and autophagy via TGF-β1/Smad3 and p38/PPARα pathways.
Collapse
|
566
|
Wang HH, Huang JH, Sue MH, Ho WC, Hsu YH, Chang KC, Chang MS. Interleukin-24 protects against liver injury in mouse models. EBioMedicine 2021; 64:103213. [PMID: 33508745 PMCID: PMC7841303 DOI: 10.1016/j.ebiom.2021.103213] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/11/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Background Interleukin-24 (IL-24) binds to two kinds of receptor complexes, namely IL-20R1/IL-20R2 and IL-20R2/IL-22R1, which are also bound by IL-20. IL-20 plays a detrimental role in liver fibrosis. Due to the sharing of receptor complexes, we aimed to determine whether IL-24 also participates in liver fibrosis. Methods Clinical biopsy specimens from various stages of liver fibrosis were used to analyze IL-24 expression. IL-24 protein was administered to mice with thioacetamide (TAA)-induced liver injury. The direct effects of IL-24 on mouse primary hepatocytes or hepatic stellate cells (HSCs) were analyzed. Wild-type, IL-20R1-, and IL20R2-deficient mice were used to establish a model of acute TAA-induced liver injury. Findings Among patients with more severe liver fibrosis, there was a reduced IL-24/IL-20 ratio. Administration of IL-24 protein protected mice from TAA-induced liver injury and reduction of liver inflammation by antioxidant effects. IL-24 protected hepatocytes from TAA-induced apoptosis and prevented liver fibrosis through the inhibition of the HSCs activation. The protective role of IL-24 acted on liver cells were mainly IL-20R1-independent. IL-20R2-deficient mice exhibited more severe liver injury upon TAA treatment, thus confirming the protective role of IL-24. Interpretation IL-24 plays a key protective role in the progression of liver injury and has therapeutic potential for treating liver injuries. Funding This work was supported by the Ministry of Science and Technology of Taiwan (MOST 106–2320-B-006–024) and Taiwan Liver Disease Prevention & Treatment Research Foundation.
Collapse
Affiliation(s)
- Hsiao-Hsuan Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jian-Hao Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Min-Hao Sue
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Chih Ho
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hsiang Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Kung-Chao Chang
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Shi Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
567
|
Fang H, Yu L, You D, Peng N, Guo W, Wang J, Zhang X. In vivo Therapeutic Effects and Mechanisms of Hydroxyasiaticoside Combined With Praziquantel in the Treatment of Schistosomiasis Induced Hepatic Fibrosis. Front Bioeng Biotechnol 2021; 8:613784. [PMID: 33553120 PMCID: PMC7862569 DOI: 10.3389/fbioe.2020.613784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Schistosomiasis has been a fatal obstinate disease that threatens global human health, resulting in the granulomatous inflammation and liver fibrosis. Objective:The aim of this study was to evaluate the therapeutic effects and mechanisms of hydroxyasiaticoside combined with praziquantel in the treatment of schistosomiasis-induced liver fibrosis. Methods:Mice were randomly distributed into four experimental groups: normal control group, model group, praziquantel group, praziquantel + hydroxyasiaticoside group. Except for the normal control group, they were infected with Schistosomia cercariae through the abdominal skin to induce liver fibrosis. In the intervention group, mice were administered with the respective drugs by gavage after 8 weeks of infection. At the end of the treatment, mice were sacrificed to collect blood for the determination of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) serum levels. Moreover, the liver was excised, weighed, and liver indices were calculated. Histopathological examination was performed to assess liver morphology. Besides, the expression of collagen type I and III in liver was determined; the mRNA expression levels of IL-6 and TNF-α in liver tissues were measured using Real-time PCR while ELISA and western blotting were performed on liver tissue homogenate to determine the protein expression of IL-6 and TNF-α. Results:The combination of praziquantel and hydroxyasiaticoside lowered the pathological scores of schistosomiasis-induced hepatic fibrosis, the liver indice, serum AST and ALT levels, improved liver morphology, downregulated the expression levels of hepatic type I and III collagen, inhibited the mRNA expression levels of pro-inflammatory factors (IL-6 and TNF-α) in the liver of mice relative to the praziquantel alone. Conclusion:The combination of hydroxyasiaticoside and praziquantel is a potential therapeutic option for schistosomiasis-induced hepatic fibrosis. Notably, this combination noticeably suppresses the protein and mRNA expression levels of pro-inflammatory factors (TNF-α and IL-6) in the liver.
Collapse
Affiliation(s)
- Huilong Fang
- Department of Pharmacology, Xiangnan University, Chenzhou, China
| | - Ling Yu
- Affiliated Hospital of Xiangnan University, Chenzhou, China
| | - Da You
- Department of Pharmacology, Xiangnan University, Chenzhou, China
| | - Nan Peng
- Department of Pharmacology, Xiangnan University, Chenzhou, China
| | - Wanbei Guo
- Department of Pharmacology, Xiangnan University, Chenzhou, China
| | - Junjie Wang
- Department of Pharmacology, Xiangnan University, Chenzhou, China
| | - Xing Zhang
- Department of Trauma and Reconstructive Surgery, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen, Germany
| |
Collapse
|
568
|
An L, Lin Y, Li L, Kong M, Lou Y, Wu J, Liu Z. Integrating Network Pharmacology and Experimental Validation to Investigate the Effects and Mechanism of Astragalus Flavonoids Against Hepatic Fibrosis. Front Pharmacol 2021; 11:618262. [PMID: 33551818 PMCID: PMC7862122 DOI: 10.3389/fphar.2020.618262] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 01/18/2023] Open
Abstract
Hepatic fibrosis (HF) represents the excessive wound healing where an excess amount of connective tissues is formed within the liver, finally resulting in cirrhosis or even hepatocellular carcinoma (HCC). Therefore, it is significant to discover the efficient agents and components to treat HF, thus restraining the further progression of hepatopathy. Astragalus membranaceus (Fisch.) Bunge [also called Astragali Radix (AR)] is a famous herb in traditional Chinese medicine (TCM), which possesses a variety of biological activities and exerts good therapeutic effects in the treatment of HF. Flavonoids account for the major active ingredients related to the AR pharmacological effects. Total AR flavonoids have been proved to exert inhibitory effects on hepatic fibrosis. This study aimed to further undertake network pharmacology analysis coupled with experimental validation and molecular docking to investigate the effects and mechanism of multiple flavonoid components from AR against liver fibrosis. The results of the network pharmacology analysis showed that the flavonoids from AR exerted their pharmacological effects against liver fibrosis by modulating multiple targets and pathways. The experimental validation data showed that the flavonoids from AR were able to suppress transforming growth factor beta 1 (TGF-β1)-mediated activation of hepatic stellate cells (HSCs) and reduce extracellular matrix deposition in HSC-T6 cells via regulating the nuclear factor kappa B (NF-κB) signal transduction pathway. The results of the molecular docking study further showed that the flavonoids had a strong binding affinity for IκB kinase (IKKβ) after docking into the crystal structure. The above results indicated that, flavonoids possibly exerted the anti-inflammatory effect on treating HF by mediating inflammatory signaling pathways. The potential mechanism of these flavonoids against liver fibrosis may be related to suppression of the NF-κB pathway through effective inhibition of IKKβ. This study not only provides a scientific basis for clarifying the effects and mechanism of AR flavonoids against liver fibrosis but also suggests a novel promising therapeutic strategy for the treatment of liver fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinjun Wu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
569
|
Tian X, Zhao H, Guo Z. Effects of Carvedilol on the Expression of TLR4 and its Downstream Signaling Pathway in the Liver Tissues of Rats with Cholestatic Liver Fibrosis. Curr Mol Med 2021; 20:708-716. [PMID: 32077825 DOI: 10.2174/1566524020666200220130705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 11/22/2022]
Abstract
Objectives:
This study was designed to investigate the effects of carvedilol
on the expression of TLR4 and its downstream signaling pathway in the liver tissues of
rats with cholestatic liver fibrosis and provide experimental evidence for clinical
treatment of liver fibrosis with carvedilol.
Methods:
A total of fifty male Sprague Dawley rats were randomly divided into five
groups (10 rats per group): sham operation (SHAM) control group, bile duct ligation
(BDL) model group, low-dose carvedilol treatment group (0.1mg·kg-1·d-1), medium-dose
carvedilol treatment group (1mg·kg-1·d-1), and high-dose carvedilol treatment group
(10mg·kg-1·d-1). Rat hepatic fibrosis model was established by applying BDL. Forty-eight
hours after the operation, carvedilol was administered twice a day. The blood and liver
were simultaneously collected under the aseptic condition for further detection in two
weeks after the operation. The alanine aminotransferase (ALT), aspartate
aminotransferase (AST), total bilirubin (TBil) and albumin (Alb) in serum were measured.
HE and Masson staining were used to determine hepatic fibrosis degree. Hydroxyproline
assay was employed to detect liver collagen synthesis. Western Blot was used to
measure the expression of TLR4, NF-κB p65 and β-arrestin2 protein. Quantitative
analysis of TLR4, MyD88, TNF-α and IL-6 mRNA was performed by Realtime-PCR.
Results:
Compared with the SHAM group, the BDL group showed obvious liver injury,
increased levels of inflammatory factors, and continued progression of liver fibrosis. The
above changes in the BDL group were alleviated in the carvedilol treatment groups. The
improvement effects augmented as dosages increased. In addition, compared with the
BDL group, the reduction of the expressions of TLR4, MyD88 and NF-κB p65 in liver
tissues and the increase of the expression of β -arrestin2 in the high-dose carvedilol
group were more significant.
Conclusions:
Carvedilol can reduce the release of inflammatory mediators by downregulating
TLR4 expression and inhibiting its downstream signaling pathway, thus
playing a potential therapeutic role in cholestatic liver fibrosis.
Collapse
Affiliation(s)
- Xiaopeng Tian
- Department of Gastroenterology, Xingtai People’s Hospital, Xingtai, Hebei 054000, China
| | - Huimin Zhao
- Department of Gastroenterology, Xingtai People’s Hospital, Xingtai, Hebei 054000, China
| | - Zengcai Guo
- Department of Gastroenterology, Xingtai People’s Hospital, Xingtai, Hebei 054000, China
| |
Collapse
|
570
|
Keshavarz Azizi Raftar S, Abdollahiyan S, Azimirad M, Yadegar A, Vaziri F, Moshiri A, Siadat SD, Zali MR. The Anti-fibrotic Effects of Heat-Killed Akkermansia muciniphila MucT on Liver Fibrosis Markers and Activation of Hepatic Stellate Cells. Probiotics Antimicrob Proteins 2021; 13:776-787. [PMID: 33433897 DOI: 10.1007/s12602-020-09733-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 12/17/2022]
Abstract
Hepatic stellate cell (HSC) activation is a key phenomenon in development of liver fibrosis. Recently, Akkermansia muciniphila has been introduced as a next-generation microbe residing in the mucosal layer of the human gut. Due to the probable risks associated with the use of live probiotics, the tendency to use heat-killed bacteria has been raised. Herein, we investigated the potential anti-fibrotic effects of heat-killed A. muciniphila MucT on activation of HSCs. The human LX-2 cells were stimulated by various concentrations of LPS to evaluate the optimal concentration for HSC activation. Cell viability of LX-2 cells treated with LPS and heat-killed A. muciniphila MucT was measured by MTT assay. Scanning electron microscopy was used to analyze the morphology of heat-killed bacteria. Quiescent and LPS-stimulated LX-2 cells were coinfected with heat-killed A. muciniphila MucT. The gene expression of α-SMA, TIMP, Col1, TGF-β, TLR4, and PPARγ was analyzed using quantitative real-time PCR. Our results showed that LPS treatment led to a significant increase in fibrosis markers in a concentration-independent manner (P < 0.0001), and significantly downregulated the expression of PPARγ (P < 0.0001). The heat-killed A. muciniphila MucT could significantly modulate the expression of fibrosis markers particularly in MOI 10 (P < 0.0001), and reversed the HSC activation in LPS-stimulated LX-2 cells. In conclusion, we demonstrated that heat-killed A. muciniphila MucT was safe and capable to ameliorate LPS-induced HSC activation through modulation of fibrosis markers. Further in vivo studies are required to validate the anti-fibrotic properties of heat-killed A. muciniphila MucT.
Collapse
Affiliation(s)
- Shahrbanoo Keshavarz Azizi Raftar
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran.,Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Abdollahiyan
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farzam Vaziri
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Arfa Moshiri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Experimental Therapy Unit, Laboratory of Oncology, G. Gaslini Children's Hospital, Genoa, Italy
| | - Seyed Davar Siadat
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Mycobacteriology and Pulmonary Research Department, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
571
|
Guo J, Liu W, Zeng Z, Lin J, Zhang X, Chen L. Tgfb3 and Mmp13 regulated the initiation of liver fibrosis progression as dynamic network biomarkers. J Cell Mol Med 2021; 25:867-879. [PMID: 33269546 PMCID: PMC7812286 DOI: 10.1111/jcmm.16140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/29/2020] [Accepted: 11/13/2020] [Indexed: 01/18/2023] Open
Abstract
Liver fibrogenesis is a complex scar-forming process in the liver. We suggested that the liver first responded to chronic injuries with gradual changes, then reached the critical state and ultimately resulted in cirrhosis rapidly. This study aimed to identify the tipping point and key molecules driving liver fibrosis progression. Mice model of liver fibrosis was induced by thioacetamide (TAA), and liver tissues were collected at different time-points post-TAA administration. By dynamic network biomarker (DNB) analysis on the time series of liver transcriptomes, the week 9 post-TAA treatment (pathologically relevant to bridging fibrosis) was identified as the tipping point just before the significant fibrosis transition, with 153 DNB genes as key driving factors. The DNB genes were functionally enriched in fibrosis-associated pathways, in particular, in the top-ranked DNB genes, Tgfb3 negatively regulated Mmp13 in the interaction path and they formed a bistable switching system from a dynamical perspective. In the in vitro study, Tgfb3 promoted fibrogenic genes and down-regulate Mmp13 gene transcription in an immortalized mouse HSC line JS1 and a human HSC line LX-2. The presence of a tipping point during liver fibrogenesis driven by DNB genes marks not only the initiation of significant fibrogenesis but also the repression of the scar resolution.
Collapse
Affiliation(s)
- Jinsheng Guo
- Department of Gastroenterology and HepatologyZhong Shan HospitalFu Dan UniversityShanghai Institute of Liver DiseasesShanghaiChina
| | - Weixin Liu
- Key Laboratory of Systems BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
| | - Zhiping Zeng
- Department of Gastroenterology and HepatologyZhong Shan HospitalFu Dan UniversityShanghai Institute of Liver DiseasesShanghaiChina
| | - Jie Lin
- Key Laboratory of Systems BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
| | - Xingxin Zhang
- Department of Gastroenterology and HepatologyZhong Shan HospitalFu Dan UniversityShanghai Institute of Liver DiseasesShanghaiChina
| | - Luonan Chen
- Key Laboratory of Systems BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
- Key Laboratory of Systems BiologyHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesChinese Academy of SciencesHangzhouChina
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
| |
Collapse
|
572
|
Liao J, Zhang Z, Yuan Q, Liu Q, Kuang J, Fang Y, Hu X. A lncRNA Gpr137b-ps/miR-200a-3p/CXCL14 axis modulates hepatic stellate cell (HSC) activation. Toxicol Lett 2021; 336:21-31. [PMID: 33069761 DOI: 10.1016/j.toxlet.2020.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/31/2020] [Accepted: 10/02/2020] [Indexed: 01/08/2023]
Abstract
Hepatic fibrosis is the wound healing response upon the liver tissue damage caused by multiple stimuli. Targeting activated hepatic stellate cells (HSCs), the major extracellular matrix (ECM)-producing cells within the damaged liver, has been regarded as one of the main treatments for hepatic fibrosis. In the present study, we performed preliminary bioinformatics analysis attempting to identify possible factors related to hepatic fibrosis and found that lncRNA G protein-coupled receptor 137B (Gpr137b-ps) and C-X-C motif chemokine ligand 14 (CXCL14) showed to be markedly upregulated within carbon tetrachloride (CCl4)-caused hepatic fibrotic mice tissue samples and activated HSCs. CXCL14 The silencing of lncRNA Gpr137b-ps or CXCL14 alone could significantly improve CCl4-induced fibrotic changes in mice liver in vivo and collagen I and III release by HSCs and HSC proliferation in vitro. miR-200a-3p directly targeted lncRNA Gpr137b-ps and CXCL14, respectively. LncRNA Gpr137b-ps relieved miR-200a-3p-induced inhibition on CXCL14 expression via acting as a ceRNA. In HSCs, the effects of lncRNA Gpr137b-ps silencing on collagen I and III release by HSCs and HSC proliferation were significantly reversed by miR-200a-3p inhibition, and the effects of miR-200a-3p inhibition were reversed by CXCL14 silencing. In conclusion, we demonstrated a lncRNA Gpr137b-ps/miR-200a-3p/CXCL14 axis that modulates HSC activation and might exert an effect on the pathogenesis of liver fibrosis.
Collapse
Affiliation(s)
- Jinmao Liao
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Zheng Zhang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Qi Yuan
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Qiong Liu
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Jia Kuang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Yuan Fang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Xiaoxuan Hu
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China.
| |
Collapse
|
573
|
Zhang L, Pan X, Xu L, Zhang L, Huang H. Mitochondria-targeted curcumin loaded CTPP–PEG–PCL self-assembled micelles for improving liver fibrosis therapy. RSC Adv 2021; 11:5348-5360. [PMID: 35423083 PMCID: PMC8694685 DOI: 10.1039/d0ra09589c] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Liver fibrosis, originating from activated hepatic stellate cells (HSCs), is receiving much attention in the treatment of clinical liver disease. In this study, mitochondria-targeted curcumin (Cur) loaded 3-carboxypropyl-triphenylphosphonium bromide–poly(ethylene glycol)–poly(ε-caprolactone) (CTPP–PEG–PCL) micelles were constructed to prolong the systemic circulation of Cur, improve the bioavailability of Cur and play a precise role in anti-fibrosis. The prepared Cur–CTPP–PEG–PCL micelles with a spherical shape had satisfactory dispersion, low critical micelle concentration (CMC) and high encapsulation efficiency (92.88%). The CTPP modification endowed good endosomal escape ability to the CTPP–PEG–PCL micelles, and micelles could be selectively accumulated in mitochondria, thereby inducing the enhanced cell proliferation inhibition of HSC-T6 cells. Mitochondrial Membrane Potential (MMP) was greatly reduced due to the mitochondrial-targeting of Cur. Moreover, the system circulation of Cur was extended and bioavailability was significantly enhanced in vivo. As expected, Cur loaded CTPP–PEG–PCL micelles were more effective in improving liver fibrosis compared with Cur and Cur–mPEG–PCL micelles. In conclusion, the Cur–CTPP–PEG–PCL based micelles can be a potential candidate for liver fibrosis treatment in future clinical applications. A mitochondria-targeting micelle system based on CTPP–PEG–PCL polymer was designed to deliver curcumin to active HSC-T6 cells and prolong the systemic circulation and bioavailability of curcumin in vivo for effective treatment of liver fibrosis.![]()
Collapse
Affiliation(s)
- Liqiao Zhang
- Department of Pharmacy
- Chengdu Women's and Children's Central Hospital
- School of Medicine
- University of Electronic Science and Technology of China
- Chengdu 611731
| | - Xiuhua Pan
- Key Laboratory of Modern Chinese Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Lixing Xu
- College of Pharmacy
- Nantong University
- Nantong 226001
- PR China
| | - Linlin Zhang
- Nanjing Chia Tai Tianqing Pharmaceutical CO., Ltd
- Nanjing 210000
- PR China
| | - Haiqin Huang
- College of Pharmacy
- Nantong University
- Nantong 226001
- PR China
| |
Collapse
|
574
|
Wang T, Zhou X, Kuang G, Jiang R, Guo X, Wu S, Wan J, Yin L. Paeoniflorin modulates oxidative stress, inflammation and hepatic stellate cells activation to alleviate CCl4-induced hepatic fibrosis by upregulation of heme oxygenase-1 in mice. J Pharm Pharmacol 2020; 73:338-346. [PMID: 33793876 DOI: 10.1093/jpp/rgaa042] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The role of Paeoniflorin on hepatic fibrosis and the specific mechanisms has not yet been elucidated. Therefore, we explored whether Paeoniflorin exerted protective effects on carbon tetrachloride (CCl4)-induced hepatic fibrosis and the underlying mechanisms. METHODS A model of hepatic fibrosis was induced by intraperitoneally injecting with CCl4 (10% 5 μl/g) twice a week for 7 weeks. To explore the effects of Paeoniflorin, mice were treated with Paeoniflorin (100 mg/kg) by gavage once a day at 1 week after modeling until they were sacrificed. KEY FINDINGS Paeoniflorin remarkably improved liver function and histopathological changes of hepatic tissues in CCl4-induced liver injury. Besides, the serum MAO enzyme activity and hydroxyproline contents were notably decreased following the intervention of Paeoniflorin. The decreased expression of Vimentin, α-SMA, Col1a and Desmin manifested the inhibition of the hepatic stellate cells (HSCs) activation. Interestingly, Paeoniflorin intervention significantly upregulated the expression of heme oxygenase-1, and attenuated the inflammatory cytokines production as well as the CCl4-induced oxidative stress imbalance. CONCLUSIONS Paeoniflorin could effectively alleviate CCl4-induced hepatic fibrosis by upregulation of heme oxygenase-1, and it might be a new effective option for the comprehensive treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Ting Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xu Zhou
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Ge Kuang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Xinyi Guo
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Shengwang Wu
- Department of Pharmacology, Chongqing Medical University, Chongqing, China
| | - Jingyuan Wan
- Department of Orthopaedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangjun Yin
- Department of Orthopaedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
575
|
The Effect of Lecithins Coupled Decorin Nanoliposomes on Treatment of Carbon Tetrachloride-Induced Liver Fibrosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8815904. [PMID: 33415158 PMCID: PMC7752282 DOI: 10.1155/2020/8815904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/26/2020] [Accepted: 11/27/2020] [Indexed: 02/08/2023]
Abstract
This study aimed to investigate the effect of bile duct-targeting lecithins- (PC-) coupled decorin (DCN) (PC-DCN) nanoliposomes against liver fibrosis in vitro and in vivo. We prepared PC-DCN nanoliposomes by using rat astrocytes, HSC-T6, to verify the antifibrosis effect of PC-DCN in vitro. First, we established a rat model of carbon tetrachloride-induced fibrosis. PC-DCN nanoliposomes were then injected into fibrotic rats via the portal vein or bile duct. The EdU assay was performed to analyze cell proliferation. Immunofluorescence staining was used to detect α-smooth muscle actin (α-SMA) expression. Western blot was performed to examine the expression of α-SMA, collagen type I alpha 1 (COL1A1), and transforming growth factor-β (TGF-β) protein. The levels of aspartate transaminase (AST), alanine transaminase (ALT), and total bilirubin (TBIL) were examined by enzyme-linked immunosorbent assay (ELISA) analysis. Hematoxylin and eosin (H&E) staining and Masson trichrome staining were used to determine liver tissue lesions and liver fibrosis. Compared with TGF-β group, PC-DCN treatment could significantly reduce cell proliferation. Western blot analysis indicated that the expression of α-SMA, COL1A1, and TGF-β was downregulated after treatment with PC-DCN in vitro and in vivo. Immunofluorescence staining confirmed that α-SMA expression was reduced by PC-DCN. Furthermore, H&E staining and Masson trichrome staining showed that the administration of PC-DCN nanoliposomes via the bile duct could reduce the extent of liver fibrosis. PCR analysis showed that PC-DCN administration could reduce proinflammatory cytokines IL-6, TNF-α, and IL-1β expression via the bile duct. The administration of PC-DCN nanoliposomes also significantly downregulated liver function indicators ALT, AST, and TBIL. The results of our study indicated that PC-DCN could effectively reduce the extent of liver fibrosis.
Collapse
|
576
|
Wang Q, Liu H, Zhu Z, Sheng Y, Du Y, Li Y, Liu J, Zhang J, Xing W. Feasibility of T1 mapping with histogram analysis for the diagnosis and staging of liver fibrosis: Preclinical results. Magn Reson Imaging 2020; 76:79-86. [PMID: 33242591 DOI: 10.1016/j.mri.2020.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/10/2020] [Accepted: 11/14/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE To compare the diagnostic accuracy of parameters derived from the histogram analysis of precontrast, 10-min hepatobiliary phase (HBP) and 20-min HBP T1 maps for staging liver fibrosis (LF). METHODS LF was induced in New Zealand white rabbits by subcutaneous injections of carbon tetrachloride for 4-16 weeks (n = 120), and 20 rabbits injected with saline served as controls. Precontrast, 10-min and 20-min HBP modified Look-Locker inversion recovery (MOLLI) T1 mapping was performed. Histogram analysis of T1 maps was performed, and the mean, median, skewness, kurtosis, entropy, inhomogeneity and 10th/25th/75th/90th percentiles of T1native, T110min and T120min were derived. Quantitative histogram parameters were compared. For significant parameters, further receiver operating characteristic (ROC) analyses were performed to evaluate the potential diagnostic performance in differentiating LF stages. RESULTS Finally, 17, 20, 21, 21 and 20 rabbits were included for the F0, F1, F2, F3, and F4 pathological grades of fibrosis, respectively. The mean/75th of T1native, entropy of T110min and entropy/mean/median/10th of T120min demonstrated a significant good correlation with the LF stage (|r| = 0.543-0.866, all P < 0.05). The 75th of T1native, entropy10min, and entropy20min were the three most reliable imaging markers in reflecting the stage of LF. The area under the ROC curve of entropy20min was larger than that of entropy10min (P < 0.05 for LF ≥ F2, ≥F3, and ≥ F4) and the 75th of T1native (P < 0.05 for LF ≥ F2 and ≥ F3) for staging LF. CONCLUSION Magnetic resonance histogram analysis of T1 maps, particularly the entropy derived from 20-min HBP T1 mapping, is promising for predicting the LF stage.
Collapse
Affiliation(s)
- Qing Wang
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People's Hospital, Jiangsu 213200, China.
| | - HaiFeng Liu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People's Hospital, Jiangsu 213200, China
| | - ZuHui Zhu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People's Hospital, Jiangsu 213200, China
| | - Ye Sheng
- Department of Interventional Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People's Hospital, Changzhou, Jiangsu 213200, China
| | - YaNan Du
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People's Hospital, Jiangsu 213200, China
| | - YuFeng Li
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People's Hospital, Jiangsu 213200, China
| | - JianHong Liu
- Department of Pathology, The Third People's Hospital of Changzhou, Changzhou, Jiangsu 213200, China
| | | | - Wei Xing
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People's Hospital, Jiangsu 213200, China.
| |
Collapse
|
577
|
Sun C, Zhang H, Liu X. Emerging role of CCN family proteins in fibrosis. J Cell Physiol 2020; 236:4195-4206. [PMID: 33222181 DOI: 10.1002/jcp.30171] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/30/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
Fibrosis is a common pathological change characterized by the excessive accumulation of fibrous connective tissue. Once uncontrolled, this pathological progress can lead to irreversible damage to the structure and function of organs, which is a serious threat to human health and life. Actually, the disability and death of patients caused by many chronic diseases have a closed relationship with fibrosis. The CCN protein family, including six members, is a small group of matrix proteins exhibiting structurally similar features. In the past 20 years, different biological functions of CCN proteins have been identified in various diseases. Of note, it has been recently shown that they are implicated in the key pathological process of fibrosis. In this review, we summarize the current status of knowledge regarding the role of CCN proteins involved in the pathogenesis of fibrosis diseases in detail. Furthermore, we highlight some of the underlying interaction mechanisms of CCN protein acting in fibrosis that helps to develop new drugs and determine appropriate clinical strategies for fibrotic diseases.
Collapse
Affiliation(s)
- Chao Sun
- Department of Spine Surgery, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Han Zhang
- Department of Spine Surgery, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xinhui Liu
- Department of Spine Surgery, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
578
|
Li Y, Jin C, Shen M, Wang Z, Tan S, Chen A, Wang S, Shao J, Zhang F, Zhang Z, Zheng S. Iron regulatory protein 2 is required for artemether -mediated anti-hepatic fibrosis through ferroptosis pathway. Free Radic Biol Med 2020; 160:845-859. [PMID: 32947011 DOI: 10.1016/j.freeradbiomed.2020.09.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/25/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Currently, the existing treatments have not cured the liver fibrosis thoroughly. Ferroptosis is a newly discovered way of cell death, which is closely related to many diseases. Previous studies have shown that ferroptosis plays an important role in the occurrence and development of liver fibrosis, but the further mechanism remains to be discovered. METHODS LX-2 cells were used as the research object, fibrosis activation index was detected by Western blot, PCR and Immunofluorescence, ferroptosis was detected by kits, the binding and interaction between IRP2 (iron regulatory protein 2) and STUB1 (STIP1 homology and U-box containing protein 1) were detected by Immunoprecipitation and ubiquitin test, and IRP2 knockdown mice were constructed by interfering plasmid to verify the results of in vitro experiment. RESULT Our research showed that ART (artemether) had a good anti-fibrosis effect in vivo and in vitro, and ferroptosis played an important role in this process. Further studies have found that ART could lead to the accumulation of IRP 2 a in hepatic stellate cell by inhibiting the ubiquitination of it, thus inducing the increase of iron in HSC (hepatic stellate cell), which could product a large number of ROS (reactive oxide species), resulting the occurrence of ferroptosis in cells. Our findings provided an experimental basis for ART to become a drug for the treatment of liver fibrosis. CONCLUSION Our results show that IRP2-Iron-ROS axis is necessary for ART to induce ferroptosis in HSC and play an anti-fibrotic effect.
Collapse
Affiliation(s)
- Yujia Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chun Jin
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenyi Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shanzhong Tan
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St Louis, USA
| | - Shijun Wang
- Shandong Co-innovation Center of TCM Formula, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
579
|
McQuitty CE, Williams R, Chokshi S, Urbani L. Immunomodulatory Role of the Extracellular Matrix Within the Liver Disease Microenvironment. Front Immunol 2020; 11:574276. [PMID: 33262757 PMCID: PMC7686550 DOI: 10.3389/fimmu.2020.574276] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease when accompanied by underlying fibrosis, is characterized by an accumulation of extracellular matrix (ECM) proteins and chronic inflammation. Although traditionally considered as a passive and largely architectural structure, the ECM is now being recognized as a source of potent damage-associated molecular pattern (DAMP)s with immune-active peptides and domains. In parallel, the ECM anchors a range of cytokines, chemokines and growth factors, all of which are capable of modulating immune responses. A growing body of evidence shows that ECM proteins themselves are capable of modulating immunity either directly via ligation with immune cell receptors including integrins and TLRs, or indirectly through release of immunoactive molecules such as cytokines which are stored within the ECM structure. Notably, ECM deposition and remodeling during injury and fibrosis can result in release or formation of ECM-DAMPs within the tissue, which can promote local inflammatory immune response and chemotactic immune cell recruitment and inflammation. It is well described that the ECM and immune response are interlinked and mutually participate in driving fibrosis, although their precise interactions in the context of chronic liver disease are poorly understood. This review aims to describe the known pro-/anti-inflammatory and fibrogenic properties of ECM proteins and DAMPs, with particular reference to the immunomodulatory properties of the ECM in the context of chronic liver disease. Finally, we discuss the importance of developing novel biotechnological platforms based on decellularized ECM-scaffolds, which provide opportunities to directly explore liver ECM-immune cell interactions in greater detail.
Collapse
Affiliation(s)
- Claire E. McQuitty
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Roger Williams
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Shilpa Chokshi
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Luca Urbani
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
580
|
Damiris K, Tafesh ZH, Pyrsopoulos N. Efficacy and safety of anti-hepatic fibrosis drugs. World J Gastroenterol 2020; 26:6304-6321. [PMID: 33244194 PMCID: PMC7656211 DOI: 10.3748/wjg.v26.i41.6304] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/30/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Recent progress in our understanding of the pathways linked to progression from hepatic insult to cirrhosis has led to numerous novel therapies being investigated as potential cures and inhibitors of hepatic fibrogenesis. Liver cirrhosis is the final result of prolonged fibrosis, which is an intimate balance between fibrogenesis and fibrinolysis. A number of these complex mechanisms are shared across the various etiologies of liver disease. Thankfully, investigation has yielded some promising results in regard to reversal of fibrosis, particularly the indirect benefits associated with antiviral therapy for the treatment of hepatitis B and C and the farnesoid receptor agonist for the treatment of primary biliary cholangitis and metabolic associated fatty liver disease. A majority of current clinical research is focused on targeting metabolic associated fatty liver disease and its progression to metabolic steatohepatitis and ultimately cirrhosis, with some hope of potential standardized therapeutics in the near future. With our ever-evolving understanding of the underlying pathophysiology, these therapeutics focus on either controlling the primary disease (the initial trigger of fibrogenesis), interrupting receptor ligand interactions and other intracellular communications, inhibiting fibrogenesis, or even promoting resolution of fibrosis. It is imperative to thoroughly test these potential therapies with the rigorous standards of clinical therapeutic trials in order to ensure the highest standards of patient safety. In this article we will briefly review the key pathophysiological pathways that lead to liver fibrosis and present current clinical and experimental evidence that has shown reversibility of liver fibrosis and cirrhosis, while commenting on therapeutic safety.
Collapse
Affiliation(s)
- Konstantinos Damiris
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ 07103, United States
| | - Zaid H Tafesh
- Medicine-Gastroenterology and Hepatology, Rutgers-New Jersey Medical School, Newark, NJ 07103, United States
| | - Nikolaos Pyrsopoulos
- Medicine-Gastroenterology and Hepatology, Rutgers-New Jersey Medical School, Newark, NJ 07103, United States
| |
Collapse
|
581
|
Huang H, Wang K, Liu Q, Ji F, Zhou H, Fang S, Zhu J. The Active Constituent From Gynostemma Pentaphyllum Prevents Liver Fibrosis Through Regulation of the TGF-β1/NDRG2/MAPK Axis. Front Genet 2020; 11:594824. [PMID: 33329740 PMCID: PMC7672159 DOI: 10.3389/fgene.2020.594824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022] Open
Abstract
Liver fibrosis resulting from chronic liver damage constitutes a major health care burden worldwide; however, no antifibrogenic agents are currently available. Our previous study reported that the small molecule NPLC0393 extracted from the herb Gynostemma pentaphyllum exerts efficient antifibrotic effects both in vivo and in vitro. In this study, a TMT-based quantitative proteomic study using a carbon tetrachloride (CCl4)-induced mouse model of liver fibrosis was performed to identify the potential target of NPLC0393. Combining this study with bioinformatic analysis of differentially expressed proteins between the CCl4 model and NPLC0393 treatment groups, we focused on the function of N-myc downstream-regulated gene 2 (NDRG2) involved in cell differentiation. In vitro studies showed that NPLC0393 prevented the TGF-β1 stimulation-induced decrease in the NDRG2 level in hepatic stellate cells (HSCs). Functional studies indicated that NDRG2 can inhibit the activation of HSCs by preventing the phosphorylation of ERK and JNK. Furthermore, knockdown of NDRG2 abolished the ability of NPLC0393 to inhibit HSC activation. In conclusion, these results provide information on the mechanism underlying the antifibrotic effect of NPLC0393 and shed new light on the potential therapeutic function of the TGF-β1/NDRG2/MAPK signaling axis in liver fibrosis.
Collapse
Affiliation(s)
- Hui Huang
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Kuifeng Wang
- Department of Infectious Diseases, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, China.,Suzhou GenHouse Pharmaceutical Co., Ltd., Suzhou, China
| | - Qian Liu
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Feihong Ji
- Department of Infectious Diseases, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, China.,Suzhou GenHouse Pharmaceutical Co., Ltd., Suzhou, China
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shanhua Fang
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiansheng Zhu
- Department of Infectious Diseases, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, China
| |
Collapse
|
582
|
Qiu Y, Wang Z, Zhang X, Huang P, Zhang W, Zhang K, Wang S, He L, Guo Y, Xiang A, Zhang C, Hao Q, Li M, Li W, Zhang Y. A long-acting isomer of Ac-SDKP attenuates pulmonary fibrosis through SRPK1-mediated PI3K/AKT and Smad2 pathway inhibition. IUBMB Life 2020; 72:2611-2626. [PMID: 33135306 DOI: 10.1002/iub.2389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 01/12/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, life-threatening lung disease with a poor prognosis. N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a critical negative regulator of fibrosis development. However, it's extremely short half-life greatly limits its applications. Previously, we reported an Ac-SDKP analog peptide in which Asp and Lys residues were replaced with D-amino acids (Ac-SDD KD P). Ac-SDD KD P exhibits better resistance to angiotensin-1-converting enzyme (ACE)-mediated degradation and a longer half-life than Ac-SDKP in rat and human sera. The objective of this study was to explore the potential application of Ac-SDD KD P for the treatment of IPF and to clarify the underlying mechanisms. We found that Ac-SDD KD P exerted similar antifibrotic effects as Ac-SDKP on human fetal lung fibroblast-1 (HFL-1) proliferation, α-smooth muscle actin (α-SMA), collagen I and collagen III expression, and Smad-2 phosphorylation in vitro. In vivo, Ac-SDD KD P exhibited significantly greater protective effects against bleomycin-induced pulmonary fibrosis than Ac-SDKP in mice. α-SMA, CD45, collagen I and collagen III expression, and Smad-2 phosphorylation were significantly decreased in the lungs of Ac-SDD KD P-treated but not Ac-SDKP-treated mice. Furthermore, a pull-down experiment was used to screen for molecules that interact with Ac-SDKP. Co-immunoprecipitation (Co-IP) and computer-based molecular docking experiments demonstrated an interaction between Ac-SDKP or Ac-SDD KD P (Ac-SDKP/Ac-SDD KD P) and serine/arginine-rich protein-specific kinase 1 (SRPK1) that caused inhibition SRPK1-mediated phosphatidylinositol-3 kinase/ serine/threonine kinase (PIK3/AKT) signaling pathway activation and Smad2 phosphorylation and thereby attenuated lung fibrosis. Our data suggest that long-acting Ac-SDD KD P may potentially be an effective drug for the treatment of pulmonary fibrosis. The interacting molecule and antifibrotic mechanism of Ac-SDKP/Ac-SDD KD P were also identified, providing an experimental and theoretical foundation for the clinical application of the drug.
Collapse
Affiliation(s)
- Yueyuan Qiu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Zhaowei Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xutao Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ping Huang
- The Brigade of Undergraduates, The Fourth Military Medical University, Xi'an, China
| | - Wangqian Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Kuo Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Shuning Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Lei He
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yanhai Guo
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - An Xiang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
583
|
MR elastography of liver: current status and future perspectives. Abdom Radiol (NY) 2020; 45:3444-3462. [PMID: 32705312 DOI: 10.1007/s00261-020-02656-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 02/08/2023]
Abstract
Non-invasive evaluation of liver fibrosis has evolved over the last couple of decades. Currently, elastography techniques are the most widely used non-invasive methods for clinical evaluation of chronic liver disease (CLD). MR elastography (MRE) of the liver has been used in the clinical practice for nearly a decade and continues to be widely accepted for detection and staging of liver fibrosis. With MRE, one can directly visualize propagating shear waves through the liver and an inversion algorithm in the scanner automatically converts the shear wave properties into an elastogram (stiffness map) on which liver stiffness can be calculated. The commonly used MRE method, two-dimensional gradient recalled echo (2D-GRE) sequence has produced excellent results in the evaluation of liver fibrosis in CLD from various etiologies and newer clinical indications continue to emerge. Advances in MRE technique, including 3D MRE, automated liver elasticity calculation, improvements in shear wave delivery and patient experience, are promising to provide a faster and more reliable MRE of liver. Innovations, including evaluation of mechanical parameters, such as loss modulus, displacement, and volumetric strain, are promising for comprehensive evaluation of CLD as well as understanding pathophysiology, and in differentiating various etiologies of CLD. In this review, the current status of the MRE of liver in CLD are outlined and followed by a brief description of advanced techniques and innovations in MRE of liver.
Collapse
|
584
|
Frohlich J, Kovacovicova K, Mazza T, Emma MR, Cabibi D, Foti M, Sobolewski C, Oben JA, Peyrou M, Villarroya F, Soresi M, Rezzani R, Cervello M, Bonomini F, Alisi A, Vinciguerra M. GDF11 induces mild hepatic fibrosis independent of metabolic health. Aging (Albany NY) 2020; 12:20024-20046. [PMID: 33126224 PMCID: PMC7655202 DOI: 10.18632/aging.104182] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
Background & aims: Growth Differentiation Factor 11 (GDF11) is an anti-aging factor, yet its role in liver diseases is not established. We evaluated the role of GDF11 in healthy conditions and in the transition from non-alcoholic fatty liver disease (NAFLD) to non-alcoholic steatohepatitis (NASH). Results: GDF11 mRNA levels positively correlated with NAFLD activity score and with CPT1, SREBP, PPARγ and Col1A1 mRNA levels, and associated to portal fibrosis, in morbidly obese patients with NAFLD/NASH. GDF11-treated mice showed mildly exacerbated hepatic collagen deposition, accompanied by weight loss and without changes in liver steatosis or inflammation. GDF11 triggered ALK5-dependent SMAD2/3 nuclear translocation and the pro-fibrogenic activation of HSC. Conclusions: GDF11 supplementation promotes mild liver fibrosis. Even considering its beneficial metabolic effects, caution should be taken when considering therapeutics that regulate GDF11. Methods: We analyzed liver biopsies from a cohort of 33 morbidly obese adults with NAFLD/NASH. We determined the correlations in mRNA expression levels between GDF11 and genes involved in NAFLD-to-NASH progression and with pathological features. We also exposed wild type or obese mice with NAFLD to recombinant GDF11 by daily intra-peritoneal injection and monitor the hepatic pathological changes. Finally, we analyzed GDF11-activated signaling pathways in hepatic stellate cells (HSC).
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Kristina Kovacovicova
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Maria R Emma
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Daniela Cabibi
- Department of Health Promotion Sciences, Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jude A Oben
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, United Kingdom
| | - Marion Peyrou
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of the University of Barcelona, Barcelona, Catalonia, Spain.,Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of the University of Barcelona, Barcelona, Catalonia, Spain.,Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Barcelona, Catalonia, Spain
| | - Maurizio Soresi
- Department of Health Promotion Sciences, Maternal and Infantile Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, Brescia, Italy
| | - Melchiorre Cervello
- Institute for Biomedical Research and Innovation, National Research Council (CNR), Palermo, Italy
| | - Francesca Bonomini
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Interdepartmental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, Brescia, Italy
| | - Anna Alisi
- Research Area for Multifactorial Diseases, Research Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, United Kingdom
| |
Collapse
|
585
|
Yao J, Liang X, Liu Y, Zheng M. Neddylation: A Versatile Pathway Takes on Chronic Liver Diseases. Front Med (Lausanne) 2020; 7:586881. [PMID: 33195347 PMCID: PMC7604315 DOI: 10.3389/fmed.2020.586881] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022] Open
Abstract
Neddylation is a ubiquitin-like posttranslational modification that conjugates neural precursor cell expressed developmentally downregulated-8 (Nedd8) to specific substrates for regulation of protein activity. In light of current researches, the neddylation pathway is aberrant in the pathogenesis of many diseases. In our review, we summarize the versatile roles of neddylation in chronic liver diseases (CLDs). CLDs are one of the leading causes of chronic disease-associated deaths worldwide. There are diverse etiologic agents causing CLDs, mainly including hepatitis B virus (HBV) infection, nonalcoholic fatty liver disease (NAFLD), chronic exposure to alcohol or drugs, and autoimmune causes. So far, however, there remains a paucity of effective therapeutic approach to CLDs. In this review, we summarized the role of the neddylation pathway which runs through the chronic hepatitis B/NAFLD-liver fibrosis-cirrhosis-hepatocellular carcinoma (HCC) axis, a canonical pattern in the process of CLD development and progression. The dysregulation of neddylation may provide a better understanding of CLD pathology and even a novel therapeutic strategy. Correspondingly, inhibiting neddylation via MLN4924, a small molecule compound targeting NEDD8-activating enzyme (NAE), can potently alleviate CLD progression and improve the outcome. On this basis, profiling and characterization of the neddylation pathway can provide new insights into the CLD pathology as well as novel therapeutic strategies, independently of the etiology of CLD.
Collapse
Affiliation(s)
- Jiping Yao
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xue Liang
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanning Liu
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Min Zheng
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
586
|
Ni M, Wang L, Yu H, Wen X, Yang Y, Liu G, Hu Y, Li Z. Radiomics Approaches for Predicting Liver Fibrosis With Nonenhanced T 1 -Weighted Imaging: Comparison of Different Radiomics Models. J Magn Reson Imaging 2020; 53:1080-1089. [PMID: 33043991 DOI: 10.1002/jmri.27391] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Liver fibrosis is a common process resulting from various etiologies. Sustained progression of liver fibrosis leads to cirrhosis, even hepatocellular carcinoma. Thus, noninvasive staging of liver fibrosis is of clinical importance. Radiomics is an emerging approach for staging liver fibrosis. However, the feature selection methods and classifier models are complicated, and may result in a discrepancy of diagnostic performance owing to different radiomics models. PURPOSE To identify the optimal feature selection and classifier methods for predicting liver fibrosis by using nonenhanced T1 -weighted imaging. STUDY TYPE Prospective. ANIMAL MODEL Wistar rats, total 97. FIELD STRENGTH/SEQUENCE 3T, 3D T1 -weighted images with fast-spoiled gradient echo (FSPGR). ASSESSMENT Liver fibrosis rats were induced via subcutaneous injection of a mixture of carbon tetrachloride. Rats in the control group were injected with saline. Segmentation and feature extraction were performed by 3D slicer and the image biomarker explorer (IBEX) software package. Data preprocessing, feature selection, model building, and model comparative evaluation were conducted with Python. The liver fibrosis stage was determined by pathological examination. STATISTICAL TESTS Receiver operating characteristic curve, fuzzy comprehensive evaluation. RESULTS For discriminating between F0 and F1-2, F0 and F3-4, F0 and F1-4, F0-1 and F2-4, F0-2 and F3-4, and F0-3 and F4, the accuracies of 12 radiomics models were 77.27-90.91%, 73.33-86.67%, 80.56-91.67%, 74.07-88.89%, 76.47-88.24%, and 79.49-92.31%, respectively. The AUCs of the radiomics models were 0.86-0.97, 0.85-0.95, 0.89-0.97, 0.81-0.96, 0.82-0.93, and 0.85-0.96, respectively. The least absolute shrinkage and selection operator / support vector machine (LASSO-SVM) model had high AUCs of 0.93-0.97. For discriminating between F0 and F1-2, F0 and F3-4, F0 and F1-4, F0-1 and F2-4, and F0-2 and F3-4, the fuzzy comprehensive evaluation showed that the LASSO-SVM model had a high fuzzy score/order of 0.087-0.091/1. DATA CONCLUSION LASSO-SVM appears to be the optimal model for predicting liver fibrosis by using nonenhanced T1 -weighted imaging in a rodent model of liver fibrosis. LEVEL OF EVIDENCE 2. TECHNICAL EFFICACY STAGE 2.
Collapse
Affiliation(s)
- Ming Ni
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiyang Yu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyi Wen
- Department of Statistics and Actuarial Science, University of Hong Kong, Hong Kong, China
| | - Yinghua Yang
- College of Computer Science and Engineering, Shandong University of Science and Technology, Qingdao, China
| | - Guangzhen Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yabin Hu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhiming Li
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
587
|
Papatheodoridi M, Mazza G, Pinzani M. Regenerative hepatology: In the quest for a modern prometheus? Dig Liver Dis 2020; 52:1106-1114. [PMID: 32868215 DOI: 10.1016/j.dld.2020.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Abstract
As liver-related morbidity and mortality is rising worldwide and orthotopic liver transplantation (OLT) remains the only standard-of-care for end-stage liver disease or acute liver failure, shortage of donor organs is becoming more prominent. Importantly, advances in regenerative Hepatology and liver bioengineering are bringing new hope to the possibility of restoring impaired hepatic functionality in the presence of acute or chronic liver failure. Hepatocyte transplantation and artificial liver-support systems were the first strategies used in regenerative hepatology but have presented various types of efficiency limitations restricting their widespread use. In parallel, liver bioengineering has been a rapidly developing field bringing continuously novel advancements in biomaterials, three dimensional (3D) scaffolds, cell sources and relative methodologies for creating bioengineered liver tissue. The current major task in liver bioengineering is to build small implantable liver mass for treating inherited metabolic disorders, bioengineered bile ducts for congenital biliary defects and large bioengineered liver organs for transplantation, as substitutes to donor-organs, in cases of acute or acute-on-chronic liver failure. This review aims to summarize the state-of-the-art and upcoming technologies of regenerative Hepatology that are emerging as promising alternatives to the current standard-of care in liver disease.
Collapse
Affiliation(s)
- Margarita Papatheodoridi
- Sheila Sherlock Liver Unit, Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Giuseppe Mazza
- Sheila Sherlock Liver Unit, Institute for Liver and Digestive Health, University College London, London, United Kingdom
| | - Massimo Pinzani
- Sheila Sherlock Liver Unit, Institute for Liver and Digestive Health, University College London, London, United Kingdom.
| |
Collapse
|
588
|
Mukherji A, Dachraoui M, Baumert TF. Perturbation of the circadian clock and pathogenesis of NAFLD. Metabolism 2020; 111S:154337. [PMID: 32795560 PMCID: PMC7613429 DOI: 10.1016/j.metabol.2020.154337] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/24/2020] [Accepted: 07/26/2020] [Indexed: 12/12/2022]
Abstract
All living organisms including humans, experience changes in the light exposure generated by the Earth's rotation. In anticipation of this unavoidable geo-physical variability, and to generate an appropriate biochemical response, species of many phyla, including mammals have evolved a nearly 24-hour endogenous timing device known as the circadian clock (CC), which is self-sustained, cell autonomous and is present in every cell type. At the heart of the 'clock' functioning resides the CC-oscillator, an elegantly designed transcriptional-translational feedback system. Notably, the core components of the CC-oscillator not only drive daily rhythmicity of their own synthesis, but also generate circadian phase-specific variability in the expression levels of thousands of target genes through transcriptional, post-transcriptional and post-translational mechanisms. Thereby, this 'clock'-system provides proper chronological coordination in the functioning of cells, tissues and organs. The CC governs many physiologically critical functions. Among these functions, the key role of the CC in maintaining metabolic homeostasis deserves special emphasis. Indeed, the several features of the modern lifestyle (e.g. travel-induced jet lag, rotating shift work, energy-dense food) which, force disruption of circadian rhythms have recently emerged as a major driver to global health problems like obesity, cardiovascular disease and metabolic liver disease such as non-alcoholic fatty liver disease (NAFLD). Here we review, the CC-dependent pathways in different tissues which play critical roles in mediating several critical metabolic functions under physiological conditions and discuss their impact for the development of metabolic disease with a focus on the liver.
Collapse
Affiliation(s)
- Atish Mukherji
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques INSERM, UMR_S 1110, Strasbourg, France.
| | - Mayssa Dachraoui
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques INSERM, UMR_S 1110, Strasbourg, France
| | - Thomas F Baumert
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hépatiques INSERM, UMR_S 1110, Strasbourg, France; Pôle Hépato-Digestif, Institut Hospitalo-Universitaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.
| |
Collapse
|
589
|
Zheng J, Chen S, Cai Y, Lin S, Ke S, Liu L. Insufficient nocturnal sleep was associated with a higher risk of fibrosis in patients with diabetes with metabolic associated fatty liver disease. Ther Adv Endocrinol Metab 2020; 11:2042018820947550. [PMID: 32973993 PMCID: PMC7493234 DOI: 10.1177/2042018820947550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Metabolic associated fatty liver disease (MAFLD) refers to metabolic dysfunction associated with fatty liver disease, and liver fibrosis stage is closely connected with liver-related and all-cause mortality. This study aimed to explore the association of sleep duration with liver fibrosis in the diabetic subgroup of the MAFLD population. METHODS This retrospective study analyzed 342 patients with MAFLD. Anthropometric measurements, clinical and biochemical markers, and lifestyle parameters were collected. Fibrosis was defined as fibrosis-4 ⩾1.3. Propensity score matching (PSM) was performed to match cases. Student's t-test and chi-square tests were applied for group comparisons, and binary regression models were used to explore the independent risk factors of liver fibrosis. RESULTS Among the 342 subjects, 87 (25.4%) were diagnosed with fibrosis and 255 (74.6%) without. Baseline characteristic comparisons showed differences in age and diabetes duration between the two groups, and adjustment was made by PSM. Ultimately, the fibrosis group and nonfibrosis group each had 87 patients. The fibrosis group had shorter duration of nocturnal sleep (6.77 ± 1.59 h) than the nonfibrosis group (7.77 ± 1.92 h, p < 0.001). More patients in the fibrosis group stayed up late at night (32.2% versus 14.9%, p < 0.01). Visceral adipose tissue (VAT) areas were larger in the fibrosis group than in the nonfibrosis group (p < 0.001). Glycemic profile, lipid profile, gamma-glutamyl transferase level, and serum uric acid level were not significantly different between the two groups. In the multivariate regression analysis, nocturnal sleep and VAT areas were independently associated with liver fibrosis, with odds ratios of 0.694 [95% confidence interval (CI) 0.551-0.875, p < 0.01] for nocturnal sleep and 1.031 (95% CI 1.014-1.048, p < 0.001) for VAT areas. CONCLUSION Insufficient nocturnal sleep was independently related to a higher risk of fibrosis. Sleep modification might be beneficial in promoting the health of patients with MAFLD.
Collapse
Affiliation(s)
- Jiaping Zheng
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Sijie Chen
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuqing Cai
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Su Lin
- Liver Research Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Sujie Ke
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Libin Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| |
Collapse
|
590
|
Boumil EF, Castro N, Phillips AT, Chatterton JE, McCauley SM, Wolfson AD, Shmushkovich T, Ridilla M, Bernstein AM. USP10 Targeted Self-Deliverable siRNA to Prevent Scarring in the Cornea. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:1029-1043. [PMID: 32829179 PMCID: PMC7452140 DOI: 10.1016/j.omtn.2020.07.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/17/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023]
Abstract
Ocular scarring after surgery, trauma, or infection leads to vision loss. The transparent cornea is an excellent model system to test anti-scarring therapies. Cholesterol-conjugated fully modified asymmetric small interfering RNAs (siRNAs) (self-deliverable siRNAs [sdRNAs]) are a novel modality for in vivo gene knockdown, transfecting cells and tissues without any additional formulations. Myofibroblasts are a main contributor to scarring and fibrosis. αv integrins play a central role in myofibroblast pathological adhesion, overcontraction, and transforming growth factor β (TGF-β) activation. Previously, we demonstrated that αv integrins are protected from intracellular degradation after wounding by upregulation of the deubiquitinase (DUB) ubiquitin-specific protease 10 (USP10), leading to integrin cell surface accumulation. In this study, we tested whether knockdown of USP10 with a USP10-targeting sdRNA (termed US09) will reduce scarring after wounding a rabbit cornea in vivo. The wounded corneal stroma was treated once with US09 or non-targeting control (NTC) sdRNA. At 6 weeks US09 treatment resulted in faster wound closure, limited scarring, and suppression of fibrotic markers and immune response. Specifically, fibronectin-extra domain A (EDA), collagen III, and a-smooth muscle actin (p < 0.05), CD45+ cell infiltration (p < 0.01), and apoptosis at 24 (p < 0.01) and 48 h (p < 0.05) were reduced post-wounding. Corneal thickness and cell proliferation were restored to unwounded parameters. Targeting the DUB, USP10 is a novel strategy to reduce scarring. This study indicates that ubiquitin-mediated pathways should be considered in the pathogenesis of fibrotic healing.
Collapse
Affiliation(s)
- Edward F Boumil
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Nileyma Castro
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Andrew T Phillips
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | | | | | | | | | - Marc Ridilla
- Repair Biotechnologies, 841 East Fayette Street, Syracuse, NY 13210, USA
| | - Audrey M Bernstein
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA.
| |
Collapse
|
591
|
Wang L, Wang Y, Quan J. Exosomal miR-223 derived from natural killer cells inhibits hepatic stellate cell activation by suppressing autophagy. Mol Med 2020; 26:81. [PMID: 32873229 PMCID: PMC7465359 DOI: 10.1186/s10020-020-00207-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
Background Activation of hepatic stellate cells (HSCs) is a prominent driver of liver fibrosis. We previously demonstrated that exosomes derived from natural killer (NK) cells (NK-Exo) attenuated TGF-β1-induced HSC activation. Herein, this study was designed to investigate the mechanism underlying the action of NK-Exo. Methods NK-Exo was isolated from NK-92MI cells and then administered into TGF-β1-treated LX-2 (human HSC line) cells. MiR-223 expression in NK-Exo was downregulated by transfecting NK-92MI cells with miR-223 inhibitor followed by exosome isolation. The HSC activation was evaluated by determining cell proliferation using CCK-8 assay and measuring the protein levels of α-SMA and CoL1A1 using western blot in LX-2 cells. The expression of miR-223 was detected by qRT-PCR. The interaction between miR-223 and ATG7 was analyzed by a dual-luciferase activity assay. The autophagy was evaluated by measuring the autophagy-related proteins using western blot. Results miR-223 was highly expressed in NK-Exo and inhibition of miR-223 expression in NK-Exo abrogated the inhibitory effect of NK-Exo on TGF-β-induced HSC activation. ATG7 was confirmed as a direct target of miR-223. Furthermore, treatment with the autophagy activator rapamycin and ATG7 overexpression in LX-2 cells abolished the HSC activation-suppressive effect of NK-Exo. Conclusion NK-Exo attenuated TGF-β-induced HSC activation by transferring miR-223 that inhibited autophagy via targeting ATG7.
Collapse
Affiliation(s)
- Ling Wang
- Department of Infectious Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yinghao Wang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Jun Quan
- Department of Infectious Diseases, Xiangya Hospital of Central South University, No. 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
592
|
The fibrotic response of primary liver spheroids recapitulates in vivo hepatic stellate cell activation. Biomaterials 2020; 261:120335. [PMID: 32891040 DOI: 10.1016/j.biomaterials.2020.120335] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 07/06/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023]
Abstract
A major obstacle in the development of efficient therapies for progressive liver fibrosis is the lack of representative in vitro models of liver fibrosis to aid in understanding the mechanisms of the disease and to promote the development of pharmaceuticals. Our aim was to develop a relevant in vitro mouse liver fibrosis model, based on the central hypothesis that liver fibrosis in vitro cannot be studied using only hepatic stellate cells (HSCs)-the main producer of scar tissue during fibrosis-, but requires cultures in which at least hepatocytes are integrated. We established robust methods to generate co-culture spheroids from freshly isolated mouse hepatocytes and HSCs. Characteristics and functionality of these spheroids were analyzed by qPCR of cell-type specific markers, CYP induction and immunohistochemistry. Compound toxicity was determined by ATP-assays. Hepatocytes and HSCs maintained their cell-type specific marker expression over a 15-day culture period without major hepatocyte dedifferentiation or HSC activation. Exposure of spheroids to TGFβ can directly activate HSCs, while acetaminophen exposure mounts a hepatocyte damage dependent activation of HSCs. Pharmaceuticals with known anti-fibrotic properties, such as Valproic acid and Verteporfin, reduce HSC activation in response to hepatocyte damage in these cultures. A comparison between the fibrotic response of the spheroid co-cultures and in vivo activated HSCs showed that these 3D co-cultures are more representative than the commonly used 2D HSC monocultures. Finally, we showed that the 3D cultures can be integrated in microfluidic chips. We conclude that our hepatocyte-stellate cell-spheroid cultures are a robust in vitro model of liver fibrosis. This model could be used to further unravel the mechanism of HSC activation and facilitate the discovery of, or testing for novel anti-fibrotic compounds, as these spheroids better reproduce HSC in vivo activation compared to the more traditional 2D mono-culture models.
Collapse
|
593
|
Fabris L, Cadamuro M, Cagnin S, Strazzabosco M, Gores GJ. Liver Matrix in Benign and Malignant Biliary Tract Disease. Semin Liver Dis 2020; 40:282-297. [PMID: 32162285 DOI: 10.1055/s-0040-1705109] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The extracellular matrix is a highly reactive scaffold formed by a wide array of multifunctional molecules, encompassing collagens and noncollagenous glycoproteins, proteoglycans, glycosaminoglycans, and polysaccharides. Besides outlining the tissue borders, the extracellular matrix profoundly regulates the behavior of resident cells by transducing mechanical signals, and by integrating multiple cues derived from the microenvironment. Evidence is mounting that changes in the biostructure of the extracellular matrix are instrumental for biliary repair. Following biliary damage and eventually, malignant transformation, the extracellular matrix undergoes several quantitative and qualitative modifications, which direct interactions among hepatic progenitor cells, reactive ductular cells, activated myofibroblasts and macrophages, to generate the ductular reaction. Herein, we will give an overview of the main molecular factors contributing to extracellular matrix remodeling in cholangiopathies. Then, we will discuss the structural alterations in terms of biochemical composition and physical stiffness featuring the "desmoplastic matrix" of cholangiocarcinoma along with their pro-oncogenic effects.
Collapse
Affiliation(s)
- Luca Fabris
- Department of Molecular Medicine, University of Padua, Padua, Italy.,Liver Center, Department of Medicine, Yale University, New Haven, Connecticut
| | | | - Silvia Cagnin
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mario Strazzabosco
- Liver Center, Department of Medicine, Yale University, New Haven, Connecticut
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Michigan
| |
Collapse
|
594
|
Abd Elmaaboud M, Khattab H, Shalaby S. Hepatoprotective effect of linagliptin against liver fibrosis induced by carbon tetrachloride in mice. Can J Physiol Pharmacol 2020; 99:294-302. [PMID: 32726558 DOI: 10.1139/cjpp-2020-0049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The current study aimed to investigate linagliptin for its potential role in the prevention of liver fibrosis progression. Balb-C mice were randomly allocated into five groups (10 each): (i) control; (ii) mice were injected intraperitoneally with 50 μL carbon tetrachloride (CCl4) in corn oil in a dose of 0.6 μL/g three times per week for four weeks; (iii) linagliptin was administered orally in a daily dose of 10 mg/kg simultaneously with CCl4; (iv) silymarin was administered orally in a daily dose of 200 mg/kg concomitantly with CCl4; and (v) only linagliptin was administered. Hepatic injury was manifested in the CCl4 group by elevation of biochemical parameters (alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP)), and hepatic fibrosis was evident histopathologically by increased METAVIR score and immunostaining expression of alpha-smooth muscle actin (α-SMA), as well as increased liver tissue oxidative stress parameters, transforming growth factor-β1 (TGF-β1), and mammalian target of rapamycin (mTOR). Linagliptin was able to stop the progression of liver fibrosis, evident histopathologically with reduced METAVIR score and α-SMA expression. The possible mechanism may be via suppression of oxidative stress, TGF-β1, and mTOR, which was associated with improvement of serum biochemical parameters ALT and AST. In conclusion, linagliptin might help to protect the liver against persistent injury-related consequences.
Collapse
Affiliation(s)
- Maaly Abd Elmaaboud
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Haidy Khattab
- Department of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shahinaz Shalaby
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
595
|
Metabolism of N-nitrosodimethylamine, methylation of macromolecules, and development of hepatic fibrosis in rodent models. J Mol Med (Berl) 2020; 98:1203-1213. [PMID: 32666246 DOI: 10.1007/s00109-020-01950-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 12/15/2022]
Abstract
Hepatic fibrosis and cirrhosis are chronic diseases affecting liver and a major health problem throughout the world. The hallmark of fibrosis and cirrhosis is inordinate synthesis and deposition of fibril forming collagens in the extracellular matrix of the liver leading to nodule formation and loss of normal architecture. Hepatic stellate cells play a crucial role in the pathogenesis and progression of liver fibrosis through secretion of several potent fibrogenic factors that trigger hepatocytes, portal fibrocytes, and bone marrow-derived fibroblasts to synthesize and deposit several connective tissue proteins, especially collagens between hepatocytes and space of Disse. Regulation of various events involved in the activation and transformation of hepatic stellate cells seems to be an appropriate strategy for the arrest of hepatic fibrosis and liver cirrhosis. In order to unravel the molecular mechanisms involved in the pathogenesis and progression of hepatic fibrosis, to determine proper and potent targets to arrest fibrosis, and to discover powerful therapeutic agents, a quick and reproducible animal model of hepatic fibrosis and liver cirrhosis that display all decompensating features of human condition is required. This review thoroughly evaluates the biochemical, histological, and pathological features of N-nitrosodimethylamine-induced model of liver injury, hepatic fibrosis, and early cirrhosis in rodents.
Collapse
|
596
|
Mahdinloo S, Kiaie SH, Amiri A, Hemmati S, Valizadeh H, Zakeri-Milani P. Efficient drug and gene delivery to liver fibrosis: rationale, recent advances, and perspectives. Acta Pharm Sin B 2020; 10:1279-1293. [PMID: 32874828 PMCID: PMC7451940 DOI: 10.1016/j.apsb.2020.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/22/2020] [Accepted: 02/28/2020] [Indexed: 12/17/2022] Open
Abstract
Liver fibrosis results from chronic damages together with an accumulation of extracellular matrix, and no specific medical therapy is approved for that until now. Due to liver metabolic capacity for drugs, the fragility of drugs, and the presence of insurmountable physiological obstacles in the way of targeting, the development of efficient drug delivery systems for anti-fibrotics seems vital. We have explored articles with a different perspective on liver fibrosis over the two decades, then collected and summarized the information by providing corresponding in vitro and in vivo cases. We have discussed the mechanism of hepatic fibrogenesis with different ways of fibrosis induction in animals. Furthermore, the critical chemical and herbal anti-fibrotics, biological molecules such as micro-RNAs, siRNAs, and growth factors, which can affect cell division and differentiation, are mentioned. Likewise, drug and gene delivery and therapeutic systems on in vitro and in vivo models are summarized in the data tables. This review article enlightens recent advances in emerging drugs and nanocarriers and represents perspectives on targeting strategies employed in liver fibrosis treatment.
Collapse
Affiliation(s)
- Somayeh Mahdinloo
- Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz 5166616471, Iran
| | - Seyed Hossein Kiaie
- Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz 5166616471, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Ala Amiri
- Faculty of Basic Sciences, Islamic Azad University, Science and Research Branch, Tehran 1477893855, Iran
| | - Salar Hemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 5166616471, Iran
| |
Collapse
|
597
|
Histopathological growth patterns correlate with the immunoscore in colorectal cancer liver metastasis patients after hepatectomy. Cancer Immunol Immunother 2020; 69:2623-2634. [PMID: 32601799 DOI: 10.1007/s00262-020-02632-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 06/01/2020] [Indexed: 02/06/2023]
Abstract
Various scoring systems have been proposed to predict the postoperative prognosis of colorectal liver metastasis (CRLM), including the clinical risk score (CRS), the immunoscore and so on. Recently, histopathological growth patterns (HGPs) have been recognized. However, the correlation between HGPs and the immunoscore, and their prognostic values in patients with CRLM after liver resection remain undetermined. In this study, HGPs were retrospectively evaluated in H&E-stained slides from 166 CRLM patients. The immunoscore was calculated according to the densities of immunostained CD3 + and CD8 + cells. A risk score combining HGPs, the immunoscore and the CRS was defined and divided patients into the low-, medium- and high-risk group. Our results showed that the densities of CD3 + and CD8 + cells were higher in the desmoplastic HGP (dHGP) group than in the non-dHGP group, and the proportion of high immunoscores was also higher in the dHGP group (51.9% vs. 33.0%, respectively, P = 0.020). Patients with the dHGP had significantly longer relapse-free survival (RFS) and overall survival (OS) than those with the non-HGP. The low-risk group showed significantly higher 2-year RFS and 5-year OS rates than the other two groups (RFS: 76.2%, 43.7% and 33.1%, respectively; P < 0.001; OS: 89.7%, 54.4% and 33.3%, respectively; P < 0.001). In conclusion, the dHGP correlates with relatively high immunoscores, predicting a favorable prognosis independent of the immunoscore and CRS. A novel risk score combining HGPs, the immunoscore and the CRS may be used for the stratification of CRLM patients' survival.
Collapse
|
598
|
Ye Z, Wei Y, Chen J, Yao S, Song B. Value of intravoxel incoherent motion in detecting and staging liver fibrosis: A meta-analysis. World J Gastroenterol 2020; 26:3304-3317. [PMID: 32684744 PMCID: PMC7336331 DOI: 10.3748/wjg.v26.i23.3304] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/26/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Liver fibrosis (LF) is a common pathological feature of all chronic liver diseases. With the accumulation of extracellular matrix in the fibrotic liver, true molecular water diffusion and perfusion-related diffusion are restricted. Intravoxel incoherent motion (IVIM) can capture the information on tissue diffusivity and microcapillary perfusion separately and reflect the fibrotic severity with diffusion coefficients.
AIM To investigate the diagnostic performance of IVIM in detecting and staging LF with histology as a reference standard.
METHODS A comprehensive literature search was conducted to identify studies on the diagnostic accuracy of IVIM for assessment of histologically proven LF. The stages of LF were classified as F0 (no fibrosis), F1 (portal fibrosis without septa), F2 (periportal fibrosis with few septa), F3 (septal fibrosis), and F4 (cirrhosis) according to histopathological findings. Data were extracted to calculate the pooled sensitivity, specificity, positive and negative likelihood ratios, and diagnostic odds ratio, as well as the area under the summary receiver operating characteristic curve (AUC) in each group.
RESULTS A total of 12 studies with 923 subjects were included in this meta-analysis with 5 studies (n = 465) for LF ≥ F1, 9 studies (n = 757) for LF ≥ F2, 4 studies (n = 413) for LF ≥ F3, and 6 studies (n = 562) for LF = F4. The pooled sensitivity and specificity were estimated to be 0.78 (95% confidence interval: 0.73-0.82) and 0.81 (0.74-0.86) for LF ≥ F1 detection with IVIM; 0.82 (0.79-0.86) and 0.80 (0.75-0.84) for staging F2 fibrosis; 0.85 (0.79-0.90) and 0.83 (0.77-0.87) for staging F3 fibrosis, and 0.90 (0.84-0.94) and 0.75 (0.70-0.79) for detecting F4 cirrhosis, respectively. The AUCs for LF ≥ F1, F2, F3, F4 detection were 0.862 (0.811-0.914), 0.883 (0.856-0.909), 0.886 (0.865-0.907), and 0.899 (0.866-0.932), respectively. Moderate to substantial heterogeneity was observed with inconsistency index (I2) ranging from 0% to 77.9%. No publication bias was detected.
CONCLUSION IVIM is a noninvasive tool with good diagnostic performance in detecting and staging LF. Optimized and standardized IVIM protocols are needed to further improve its diagnostic accuracy in clinical practice.
Collapse
Affiliation(s)
- Zheng Ye
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Yi Wei
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jie Chen
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Shan Yao
- West China School of Medicine, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
599
|
Lambrecht J, van Grunsven LA, Tacke F. Current and emerging pharmacotherapeutic interventions for the treatment of liver fibrosis. Expert Opin Pharmacother 2020; 21:1637-1650. [PMID: 32543284 DOI: 10.1080/14656566.2020.1774553] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Chronic liver disease is due to various causes of persistent liver damage and will eventually lead to the development of liver fibrosis. If no treatment is initiated, this condition may progress to cirrhosis and hepatocellular carcinoma. Current treatments comprise the elimination of the cause of injury, such as by lifestyle changes, alcohol abstinence, and antiviral agents. However, such etiology-driven therapy is often insufficient in patients with late-stage fibrosis/cirrhosis, therefore maintaining the need for efficient antifibrotic pharmacotherapeutic interventions. AREAS COVERED The authors discuss the recent advances in the development of antifibrotic drugs, which target various pathways of the fibrogenesis process, including cell death, inflammation, gut-liver axis, and myofibroblast activation. Due to the significant burden of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH), various agents which specifically target metabolic pathways and their related receptors/ligands have been developed. For some of them, e.g., obeticholic acid, advanced stage clinical trials indicate antifibrotic efficacy in NAFLD and NASH. EXPERT OPINION Significant advances have been made in the development of novel antifibrotic pharmacotherapeutics. The authors expect that the development of combinatorial therapies, which combine compounds that target various pathways of fibrosis progression, will have a major impact as future etiology-independent therapies.
Collapse
Affiliation(s)
- Joeri Lambrecht
- Liver Cell Biology Research Group, Vrije Universiteit Brussel , Brussels, Belgium
| | - Leo A van Grunsven
- Liver Cell Biology Research Group, Vrije Universiteit Brussel , Brussels, Belgium
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medical Center , Berlin, Germany
| |
Collapse
|
600
|
Roseotoxin B alleviates cholestatic liver fibrosis through inhibiting PDGF-B/PDGFR-β pathway in hepatic stellate cells. Cell Death Dis 2020; 11:458. [PMID: 32541811 PMCID: PMC7296008 DOI: 10.1038/s41419-020-2575-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023]
Abstract
Identifying effective anti-fibrotic therapies is a major clinical need that remains unmet. In the present study, roseotoxin B was shown to possess an improving effect on cholestatic liver fibrosis in bile duct–ligated mice, as proved by histochemical and immunohistochemical staining, hepatic biochemical parameters, and TUNEL apoptotic cell detection in tissue sections. Using cellular thermal shift assay, computational molecular docking, microscale thermophoresis technology, and surface plasmon resonance biosensor, we confirmed that PDGFR-β was a direct target of roseotoxin B in fibrotic livers. Of note, human tissue microarrays detected pathologically high expression of p-PDGFR-β in liver samples of ~80% of patients with liver fibrosis and cirrhosis. PDGF-B/PDGFR-β pathway promotes transdifferentiation and excessive proliferation of hepatic stellate cells (HSCs), which is a very crucial driver for liver fibrosis. Meaningfully, roseotoxin B blocked the formation of PDGF-BB/PDGFR-ββ complex by targeting the D2 domain of PDGFR-β, thereby inhibiting the PDGF-B/PDGFR-β pathway in HSCs. In summary, our study provided roseotoxin B as a unique candidate agent for the treatment of liver fibrosis.
Collapse
|